1
|
Regulation of the tumor immune microenvironment by cancer-derived circular RNAs. Cell Death Dis 2023; 14:132. [PMID: 36797245 PMCID: PMC9935907 DOI: 10.1038/s41419-023-05647-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/18/2023]
Abstract
Circular RNA (circRNAs) is a covalently closed circular non-coding RNA formed by reverse back-splicing from precursor messenger RNA. It is found widely in eukaryotic cells and can be released to the surrounding environment and captured by other cell types. This, circRNAs serve as connections between different cell types for the mediation of multiple signaling pathways. CircRNAs reshape the tumor microenvironment (TME), a key factor involved in all stages of cancer development, by regulating epithelial-stromal transformation, tumor vascularization, immune cell function, and inflammatory responses. Immune cells are the most abundant cellular TME components, and they have profound toxicity to cancer cells. This review summarizes circRNA regulation of immune cells, including T cells, natural killer cells, and macrophages; highlights the impact of circRNAs on tumor progression, treatment, and prognosis; and indicates new targets for tumor immunotherapy.
Collapse
|
2
|
Tools and methods for identification and analysis of rare antigen-specific T lymphocytes. EXPERIENTIA SUPPLEMENTUM (2012) 2014; 104:73-88. [PMID: 24214619 DOI: 10.1007/978-3-0348-0726-5_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
T lymphocytes are essential as effector and memory cells for immune defense against infections and as regulatory T cells in the establishment and maintenance of immune tolerance. However, they are also involved in immune pathology being effectors in autoimmune and allergic diseases or suppressors of immunity in cancer, and they often cause problems in transplantation. Therefore, strategies are being developed that allow the in vivo amplification or isolation, in vitro expansion and genetic manipulation of beneficial T cells for adoptive cell therapies or for the tolerization, or elimination of pathogenic T cells. The major goal is to make use of the exquisite antigen specificity of T cells to develop targeted strategies and to develop techniques that allow for the identification and depletion or enrichment of very often rare antigen-specific naïve as well as effector and memory T cells. Such techniques are very useful for immune monitoring of T cell responses in diagnostics and vaccination and for the development of T cell-based assays for the replacement of animal testing in immunotoxicology to identify contact allergens and drugs that cause adverse reactions.
Collapse
|
3
|
Immune monitoring in cancer vaccine clinical trials: critical issues of functional flow cytometry-based assays. BIOMED RESEARCH INTERNATIONAL 2013; 2013:726239. [PMID: 24195078 PMCID: PMC3806162 DOI: 10.1155/2013/726239] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 08/19/2013] [Indexed: 11/17/2022]
Abstract
The development of immune monitoring assays is essential to determine the immune responses against tumor-specific antigens (TSAs) and tumor-associated antigens (TAAs) and their possible correlation with clinical outcome in cancer patients receiving immunotherapies. Despite the wide range of techniques used, to date these assays have not shown consistent results among clinical trials and failed to define surrogate markers of clinical efficacy to antitumor vaccines. Multiparameter flow cytometry- (FCM-) based assays combining different phenotypic and functional markers have been developed in the past decade for informative and longitudinal analysis of polyfunctional T-cells. These technologies were designed to address the complexity and functional heterogeneity of cancer biology and cellular immunity and to define biomarkers predicting clinical response to anticancer treatment. So far, there is still a lack of standardization of some of these immunological tests. The aim of this review is to overview the latest technologies for immune monitoring and to highlight critical steps involved in some of the FCM-based cellular immune assays. In particular, our laboratory is focused on melanoma vaccine research and thus our main goal was the validation of a functional multiparameter test (FMT) combining different functional and lineage markers to be applied in clinical trials involving patients with melanoma.
Collapse
|
4
|
Edwards LJ, Zarnitsyna VI, Hood JD, Evavold BD, Zhu C. Insights into T cell recognition of antigen: significance of two-dimensional kinetic parameters. Front Immunol 2012; 3:86. [PMID: 22566966 PMCID: PMC3342060 DOI: 10.3389/fimmu.2012.00086] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 04/03/2012] [Indexed: 01/06/2023] Open
Abstract
The T cell receptor (TCR) interacts with peptide-major histocompatibility complex (pMHC) to enable T cell development and trigger adaptive immune responses. For this reason, TCR:pMHC interactions have been intensely studied for over two decades. However, the details of how various binding parameters impact T cell activation remain elusive. Most measurements were made using recombinant proteins by surface plasmon resonance, a three-dimensional (3D) technique in which fluid-phase receptors and ligands are removed from their cellular environment. This approach found TCR:pMHC interactions with relatively low affinities and slow off-rates for agonist peptides. Newer generation techniques have analyzed TCR:pMHC interactions in two dimensions (2D), with both proteins anchored in apposing plasma membranes. These approaches reveal in situ TCR:pMHC interaction kinetics that are of high affinity and exhibit rapid on- and off-rates upon interaction with agonist ligands. Importantly, 2D binding parameters correlate better with T cell functional responses to a spectrum of ligands than 3D measures.
Collapse
Affiliation(s)
- Lindsay J Edwards
- Department of Microbiology and Immunology, Emory University Atlanta, GA, USA
| | | | | | | | | |
Collapse
|
5
|
Davis MM, Altman JD, Newell EW. Interrogating the repertoire: broadening the scope of peptide-MHC multimer analysis. Nat Rev Immunol 2011; 11:551-8. [PMID: 21760610 PMCID: PMC3699324 DOI: 10.1038/nri3020] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Labelling antigen-specific T cells with peptide-MHC multimers has provided an invaluable way to monitor T cell-mediated immune responses. A number of recent developments in this technology have made these multimers much easier to make and use in large numbers. Furthermore, enrichment techniques have provided a greatly increased sensitivity that allows the analysis of the naive T cell repertoire directly. Thus, we can expect a flood of new information to emerge in the coming years.
Collapse
Affiliation(s)
- Mark M Davis
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA.
| | | | | |
Collapse
|
6
|
Grützkau A, Radbruch A. Small but mighty: How the MACS®-technology based on nanosized superparamagnetic particles has helped to analyze the immune system within the last 20 years. Cytometry A 2010; 77:643-7. [DOI: 10.1002/cyto.a.20918] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
7
|
Simultaneous detection of many T-cell specificities using combinatorial tetramer staining. Nat Methods 2009; 6:497-9. [PMID: 19543286 PMCID: PMC2731062 DOI: 10.1038/nmeth.1344] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Accepted: 05/18/2009] [Indexed: 01/06/2023]
Abstract
The direct detection of antigen-specific T cells using tetramers of soluble peptide-major histocompatibilty complex (pMHC) molecules is widely used in both basic and clinical immunology. However, the number of specificities that can be assessed simultaneously has been a major limitation. Here we describe and validate a method using combinations of fluorescent pMHC tetramers to simultaneously detect and enrich for many (>or=15) T-cell specificities in a single human blood sample.
Collapse
|
8
|
Monu N, Frey AB. Suppression of proximal T cell receptor signaling and lytic function in CD8+ tumor-infiltrating T cells. Cancer Res 2008; 67:11447-54. [PMID: 18056473 DOI: 10.1158/0008-5472.can-07-1441] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD8(+) tumor-infiltrating lymphocytes (TIL) lack in vivo and in vitro lytic function due to a signaling deficit characterized by failure to flux calcium or activate tyrosine kinase activity upon contact with cognate tumor cells. Although CD3 zeta is phosphorylated by conjugation in vitro with cognate tumor cells, showing that TIL are triggered, PLC gamma-1, LAT, and ZAP70 are not activated and LFA-1 is not affinity-matured, and because p56(lck) is required for LFA-1 activation, this implies that the signaling blockade is very proximal. Here, we show that TIL signaling defects are transient, being reversed upon purification and brief culture in vitro, implying a fast-acting "switch". Biochemical analysis of purified nonlytic TIL shows that contact with tumor cells causes transient activation of p56(lck) ( approximately 10 s) which is rapidly inactivated. In contrast, tumor-induced activation of p56(lck) in lytic TIL is sustained coincident with downstream TCR signaling and lytic function. Shp-1 is robustly active in nonlytic TIL compared with lytic TIL, colocalizes with p56(lck) in nonlytic TIL, and inhibition of Shp-1 activity in lytic TIL in vitro blocks tumor-induced defective TIL cytolysis. Collectively, our data support the notion that contact of nonlytic TIL with tumor cells, and not with tumor-infiltrating myeloid-derived suppressor cells, causes activation of Shp-1 that rapidly dephosphorylates the p56(lck) activation motif (Y394), thus inhibiting effector phase functions.
Collapse
Affiliation(s)
- Ngozi Monu
- Department of Cell Biology, New York University Cancer Institute, New York University School of Medicine, New York, New York 10016, USA
| | | |
Collapse
|
9
|
De Vries IJM, Bernsen MR, van Geloof WL, Scharenborg NM, Lesterhuis WJ, Rombout PDM, Van Muijen GNP, Figdor CG, Punt CJA, Ruiter DJ, Adema GJ. In situ detection of antigen-specific T cells in cryo-sections using MHC class I tetramers after dendritic cell vaccination of melanoma patients. Cancer Immunol Immunother 2007; 56:1667-76. [PMID: 17440724 PMCID: PMC11030669 DOI: 10.1007/s00262-007-0304-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2006] [Accepted: 02/21/2007] [Indexed: 12/22/2022]
Abstract
Application of tetrameric MHC class I-peptide complexes has significantly improved the monitoring of antigen-specific T cell immune responses in mouse models as well as in clinical studies. Especially MHC class I tetramer analysis of tumor-specific T cells in suspension or on thick vibratome sections from viable tissue has been proven extremely useful. Using the well-characterized mouse tyrosinase-related-protein-2 specific cytotoxic T cell (CTL) clone LP9, we now developed a method that allows for specific identification of T cells with MHC class I tetramers in 8 mum thick, chemically fixed cryosections. The protocol was validated in a murine influenza virus-infection model. Moreover, analysis of delayed type hypersensitivity (DTH) skin biopsies from melanoma patients vaccinated with peptide-loaded mature dendritic cells, revealed the presence and location of anti-tumor CTLs. The specificity of the CTLs detected in situ correlated with both the DTH challenge specificity and reactivity of cell suspensions derived from the same biopsies. Collectively, our data demonstrate that in situ MHC class I tetramer staining provides a valuable tool to reveal the presence and anatomical location of specific CTLs in frozen tissue following immune-based treatment strategies in cancer patients.
Collapse
Affiliation(s)
- I. J. M. De Vries
- Department of Tumor Immunology, Medical Oncology and Pediatric Hemato-Oncology, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | - M. R. Bernsen
- Department of Pathology, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | - W. L. van Geloof
- Department of Pathology, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | - N. M. Scharenborg
- Department of Tumor Immunology, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | - W. J. Lesterhuis
- Department of Medical Oncology, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | - P. D. M. Rombout
- Department of Pathology, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | - G. N. P. Van Muijen
- Department of Pathology, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | - C. G. Figdor
- Department of Tumor Immunology, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | - C. J. A. Punt
- Department of Medical Oncology, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | - D. J. Ruiter
- Department of Pathology, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | - G. J. Adema
- Department of Tumor Immunology, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
- Department of Tumor Immunology, NCMLS/278 TIL, University Medical Centre, Postbox 9101, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
10
|
Bharat A, Benshoff N, Fleming TP, Dietz JR, Gillanders WE, Mohanakumar T. Characterization of the role of CD8+T cells in breast cancer immunity following mammaglobin-A DNA vaccination using HLA-class-I tetramers. Breast Cancer Res Treat 2007; 110:453-63. [PMID: 17874294 DOI: 10.1007/s10549-007-9741-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2007] [Accepted: 08/17/2007] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Mammaglobin-A(mam-A) is expressed in over 80% of human breast tumors. We recently reported that mam-A DNA vaccination resulted in breast cancer immunity in a preclinical model. Here we investigated whether mam-A HLA-class-I tetramers could be used to monitor and define the role of CD8(+)cytotoxic T-lymphocytes(CTL) in mediating breast cancer immunity following mam-A DNA vaccination. STUDY DESIGN Mam-A DNA vaccination was performed in HLA-A2(+)huCD8(+ )transgenic mice. HLA-A2 tetramers carrying the immunodominant mamA2.1 peptide were used to monitor CD8(+)CTL. Human breast cancer colonies were developed in immunodeficient SCID-beige mice. ELISPOT was used to correlate frequency of mamA2.1 tetramer(+)CD8(+)T cells and IFN-gamma production [spots per million cells (spm)] in human subjects. RESULTS Vaccination of HLA-A2(+)huCD8(+) mice with mam-A DNA vaccine, but not empty vector, led to the expansion of mamA2.1 tetramer(+)CD8(+)T-cells in peripheral blood (<0.5% pre-vaccination compared to >2.0% post-vaccination). CD8(+)T cells from vaccinated mice specifically lysed UACC-812(HLA-A2(+)/mam-A(+), 25% lysis) but not MDA-MB-415(HLA-A2(-)/mam-A(+)) or MCF-7(HLA-A2(+)/mam-A(-)) breast cancer cells. Adoptive transfer of purified CD8(+)T cells from vaccinated mice into immunodeficient SCID-beige mice with established human breast cancer colonies led to tetramer(+)CD8(+ )T-cell infiltration with regression of UACC-812 but not MCF-7 tumors. HLA-A2(+) breast cancer patients revealed increased frequency of mamA2.1 tetramer(+)CD8(+ )T-cells compared to normal controls (2.86 +/- 0.8% vs. 0.71 +/- 0.1%, P = 0.01) that correlated with the IFN-gamma response to mamA2.1 peptide (48.1 +/- 20.9 vs. 2.9 +/- 0.8 spm, P = 0.03). CONCLUSIONS CD8(+ )T-cells are crucial in mediating breast cancer immunity following mam-A DNA vaccination. Mam-A HLA-class-I tetramers can be effectively used to monitor development of CD8(+ )T-cells following mam-A vaccination.
Collapse
Affiliation(s)
- Ankit Bharat
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, 3328 CSRB, P.O. Box 8109-3328, St. Louis, MO 63110, USA.
| | | | | | | | | | | |
Collapse
|
11
|
Meijer SL, Dols A, Jensen SM, Hu HM, Miller W, Walker E, Romero P, Fox BA, Urba WJ. Induction of Circulating Tumor-reactive CD8+ T Cells After Vaccination of Melanoma Patients With the gp100209-2M Peptide. J Immunother 2007; 30:533-43. [PMID: 17589294 DOI: 10.1097/cji.0b013e3180335b5e] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Patients with stage I-III melanoma were vaccinated with the modified HLA-A2-binding gp100(209-2M)-peptide after complete surgical resection of their primary lesion and sentinel node biopsy. Cytoplasmic interferon-gamma production by freshly thawed peripheral blood mononuclear cells (direct ex vivo analysis) or by peripheral blood mononuclear cells subjected to 1 cycle of in vitro sensitization with peptide, interleukin-2, and interleukin-15 was measured following restimulation with the modified and native gp100 peptides, and also A2gp100 melanoma cell lines. Peptide-reactive and tumor-reactive T cells were detected in 79% and 66% of selected patients, respectively. Patients could be classified into 3 groups according to their vaccine-elicited T-cell responses. One group of patients responded only to the modified peptide used for immunization, whereas another group of patients reacted to both the modified and native gp100 peptides, but not to naturally processed gp100 antigen on melanoma cells. In the third group of patients, circulating CD8 T cells recognized A2gp100 melanoma cell lines and also both the modified and native peptides. T cells with a low functional avidity, which were capable of lysing tumor cells only if tumor cells were first pulsed by the exogenous administration of native gp100(209-217) peptide were identified in most patients. These results indicate that vaccination with a modified gp100 peptide induced a heterogeneous group of gp100-specific T cells with a spectrum of functional avidities; however, high avidity, tumor-reactive T cells were detected in the majority of patients.
Collapse
Affiliation(s)
- Sybren L Meijer
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, 4805 NE Glisan Street, Portland, OR 97213, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
|
13
|
Generation of cytotoxic T lymphocytes specific for B-cell acute lymphoblastic leukemia family-shared peptides derived from immunoglobulin heavy chain framework region. Chin Med J (Engl) 2007. [DOI: 10.1097/00029330-200704020-00008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
14
|
Abstract
There is overwhelming evidence that the human immune system can keep in check the growth of autologous tumors. Yet, this phenomenon is rare and most often tumors survive striking a balance with the host's immune system. The well-documented coexistence of immune cells that can recognize cancer and their targets within the same host is reminiscent of chronic allograft rejection well-controlled by immune suppression or of a lingering tissue-specific autoimmune reaction. In this review, we argue that autologous tumor rejection represents a distinct form of tissue-specific rejection similar to acute allograft rejection or to flares of autoimmunity. Here we discuss similarities within the biology of these phenomena that may converge into a common immunological constant of rejection. The purpose is to simplify the basis of immune rejection to its bare bones critically dissecting the significance of those components proposed by experimental models as harbingers of this final outcome.
Collapse
Affiliation(s)
- Ena Wang
- Immunogenetics Section, The Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | | | | |
Collapse
|
15
|
Benigni F, Zimmermann VS, Hugues S, Caserta S, Basso V, Rivino L, Ingulli E, Malherbe L, Glaichenhaus N, Mondino A. Phenotype and homing of CD4 tumor-specific T cells is modulated by tumor bulk. THE JOURNAL OF IMMUNOLOGY 2005; 175:739-48. [PMID: 16002669 DOI: 10.4049/jimmunol.175.2.739] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Technical difficulties in tracking endogenous CD4 T lymphocytes have limited the characterization of tumor-specific CD4 T cell responses. Using fluorescent MHC class II/peptide multimers, we defined the fate of endogenous Leishmania receptor for activated C kinase (LACK)-specific CD4 T cells in mice bearing LACK-expressing TS/A tumors. LACK-specific CD44(high)CD62L(low) CD4 T cells accumulated in the draining lymph nodes and had characteristics of effector cells, secreting IL-2 and IFN-gamma upon Ag restimulation. Increased frequencies of CD44(high)CD62L(low) LACK-experienced cells were also detected in the spleen, lung, liver, and tumor itself, but not in nondraining lymph nodes, where the cells maintained a naive phenotype. The absence of systemic redistribution of LACK-specific memory T cells correlated with the presence of tumor. Indeed, LACK-specific CD4 T cells with central memory features (IL-2(+)IFN-gamma(-)CD44(high)CD62L(high) cells) accumulated in all peripheral lymph nodes of mice immunized with LACK-pulsed dendritic cells and after tumor resection. Together, our data demonstrate that although tumor-specific CD4 effector T cells producing IFN-gamma are continuously generated in the presence of tumor, central memory CD4 T cells accumulate only after tumor resection. Thus, the continuous stimulation of tumor-specific CD4 T cells in tumor-bearing mice appears to hinder the systemic accumulation of central memory CD4 T lymphocytes.
Collapse
Affiliation(s)
- Fabio Benigni
- Cancer Immunotherapy and Gene Therapy Program, S. Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Zimmermann VS, Benigni F, Mondino A. Immune surveillance and anti-tumor immune responses: an anatomical perspective. Immunol Lett 2005; 98:1-8. [PMID: 15790503 DOI: 10.1016/j.imlet.2004.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2004] [Revised: 09/06/2004] [Accepted: 09/10/2004] [Indexed: 11/15/2022]
Abstract
The development of adaptive immune responses against infectious agents relies on the initiation of antigen specific immune responses in secondary lymphoid organs and on the migration of effector cells at the site of infection. Similarly, the development of anti-tumor immunity depends on the recognition of tumor-derived antigens by specific lymphocytes in the context of the lymphoid tissues and on the re-localisation of the cells to the site of cell transformation. Here, we will review the preclinical studies, which have defined the spatial and temporal organisation of anti-tumor immunity, and discuss the implications of these findings in active immunotherapy.
Collapse
Affiliation(s)
- Valerie S Zimmermann
- Cancer Immunotherapy and Gene Therapy Program, San Raffaele Scientific Institute, DIBIT, Via Olgettina 58, 20132 Milan, Italy
| | | | | |
Collapse
|
17
|
Wang E, Panelli MC, Marincola FM. Understanding the response to immunotherapy in humans. ACTA ACUST UNITED AC 2005; 27:105-17. [PMID: 15666150 DOI: 10.1007/s00281-004-0198-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2004] [Accepted: 12/15/2004] [Indexed: 01/08/2023]
Abstract
Whether the efforts of the last decade aimed at the development of vaccines against tumor-specific antigens encountered success or failure is a matter of expectations. On the bright side, we could optimistically observe that anti-cancer-vaccines stand as an outstanding example of the successful implementation of modern biotechnology tools for the development of biologically sound therapeutics. In particular, vaccines against melanoma (the prototype model of tumor immunology in humans) can reproducibly induce cytotoxic T cell (CTL) responses exquisitely specific for cancer cells. This achievement trespasses the specificity of any other anti-cancer therapy. The skeptics, on the other end, might point out that immunization only rarely leads to cancer regression, labeling, therefore, this approach is ineffective. In our opinion this judgment stems from the naïve expectation that CTL induction is sufficient for an effective immune response. Here we propose that more needs to be understood about the mechanisms required for the induction of a therapeutically relevant immune response in humans. In particular, we will discuss the variables related to cancer heterogeneity, the weight of individual patients' polymorphism(s), the role of the T cell activation and differentiation and, finally, the complex relationship between immune and cancer cells within the tumor microenvironment.
Collapse
Affiliation(s)
- Ena Wang
- Immunogenetics Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892-1184, USA
| | | | | |
Collapse
|
18
|
Wang LX, Huang WX, Graor H, Cohen PA, Kim JA, Shu S, Plautz GE. Adoptive immunotherapy of cancer with polyclonal, 108-fold hyperexpanded, CD4+ and CD8+ T cells. J Transl Med 2004; 2:41. [PMID: 15566571 PMCID: PMC535812 DOI: 10.1186/1479-5876-2-41] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2004] [Accepted: 11/26/2004] [Indexed: 12/05/2022] Open
Abstract
T cell-mediated cancer immunotherapy is dose dependent and optimally requires participation of antigen-specific CD4+ and CD8+ T cells. Here, we isolated tumor-sensitized T cells and activated them in vitro using conditions that led to greater than 108-fold numerical hyperexpansion of either the CD4+ or CD8+ subset while retaining their capacity for in vivo therapeutic efficacy. Murine tumor-draining lymph node (TDLN) cells were segregated to purify the CD62Llow subset, or the CD4+ subset thereof. Cells were then propagated through multiple cycles of anti-CD3 activation with IL-2 + IL-7 for the CD8+ subset, or IL-7 + IL-23 for the CD4+ subset. A broad repertoire of TCR Vbeta families was maintained throughout hyperexpansion, which was similar to the starting population. Adoptive transfer of hyper-expanded CD8+ T cells eliminated established pulmonary metastases, in an immunologically specific fashion without the requirement for adjunct IL-2. Hyper-expanded CD4+ T cells cured established tumors in intracranial or subcutaneous sites that were not susceptible to CD8+ T cells alone. Because accessibility and antigen presentation within metastases varies according to anatomic site, maintenance of a broad repertoire of both CD4+ and CD8+ T effector cells will augment the overall systemic efficacy of adoptive immunotherapy.
Collapse
Affiliation(s)
- Li-Xin Wang
- Center for Surgery Research, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Wen-Xin Huang
- Center for Surgery Research, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Hallie Graor
- Dept. of General Surgery, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Peter A Cohen
- Center for Surgery Research, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Julian A Kim
- Dept. of General Surgery, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Suyu Shu
- Center for Surgery Research, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Gregory E Plautz
- Center for Surgery Research, The Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
19
|
Lustgarten J, Dominguez AL, Thoman M. Aged mice develop protective antitumor immune responses with appropriate costimulation. THE JOURNAL OF IMMUNOLOGY 2004; 173:4510-5. [PMID: 15383582 DOI: 10.4049/jimmunol.173.7.4510] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
There is a clear decrease in CD8(+) T cell effector function with aging, a loss once thought to be intrinsic to the CD8(+) T cells. Recent studies suggest, however, that this decline may be a consequence of altered stimulatory signals within the aged lymphoid microenvironment. In this study, we compared the immune responses of young and old mice against the BM-185 pre-B cell lymphoma expressing enhanced GFP (EGFP) as a surrogate tumor Ag. Young animals develop protective immune responses when immunized with BM-185-EGFP, but aged mice do not and ultimately succumb to the tumor. However, expression of CD80 (B7.1) on the BM-185-EGFP (BM-185-EGFP-CD80) results in rejection of the tumor by both young and old animals. Additionally, injection of BM-185-EGFP-CD80 cells in young mice promotes the development of long-lasting memory responses capable of rejecting BM-185 wild-type tumors. Aged animals similarly injected did not develop antitumor memory responses. Interestingly, old animals immunized with the BM-185-EGFP-CD80 cells plus injections of the agonist anti-OX40 mAb did develop long-lasting memory responses capable of rejecting the BM-185 wild-type tumors with the same vigor as the young animals. We show that old mice have the capacity to develop strong antitumor responses and protective memory responses as long as they are provided with efficient costimulation. These results have important implications for the development of vaccination strategies in the elderly, indicating that the aged T cell repertoire can be exploited for the induction of tumor immunity.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/biosynthesis
- Adjuvants, Immunologic/physiology
- Aging/immunology
- Animals
- Antibodies, Monoclonal/administration & dosage
- B7-1 Antigen/administration & dosage
- B7-1 Antigen/biosynthesis
- B7-1 Antigen/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- Cell Line, Tumor
- Graft Rejection/immunology
- Graft Rejection/physiopathology
- Green Fluorescent Proteins
- Immunity, Innate/immunology
- Immunologic Memory
- Injections, Subcutaneous
- Luminescent Proteins/administration & dosage
- Luminescent Proteins/immunology
- Lymphocyte Depletion
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/physiopathology
- Lymphoma, B-Cell/prevention & control
- Mice
- Mice, Inbred BALB C
- Neoplasm Transplantation/immunology
- Receptors, OX40
- Receptors, Tumor Necrosis Factor/immunology
- Receptors, Tumor Necrosis Factor/physiology
- T-Lymphocytes, Cytotoxic/immunology
Collapse
|
20
|
Lee PP. T-cell responses to cancer. Methods Cell Biol 2004; 75:513-32. [PMID: 15603440 DOI: 10.1016/s0091-679x(04)75021-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Affiliation(s)
- Peter P Lee
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
21
|
Speiser DE, Pittet MJ, Guillaume P, Lubenow N, Hoffman E, Cerottini JC, Romero P. Ex Vivo Analysis of Human Antigen-Specific CD8+ T-Cell Responses: Quality Assessment of Fluorescent HLA-A2 Multimer and Interferon-γ ELISPOT Assays for Patient Immune Monitoring. J Immunother 2004; 27:298-308. [PMID: 15235391 DOI: 10.1097/00002371-200407000-00006] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The authors developed a standardized approach for immune monitoring of antigen-specific CD8+ T cells within peripheral blood lymphocytes (PBLs) that combines direct ex vivo analysis of Melan-A/MART-1 and influenza-specific CD8+ T cells with HLA-A2/peptide multimers and interferon-gamma ELISPOT assays. Here the authors assessed the quality of results obtained with 180 PBLs from healthy donors and melanoma patients. Reproducibility of the multimer assay was good (average of 15% variation). In the absence of in vivo antigen-specific T-cell responses, physiologic fluctuations of multimer-positive T cells was low, with variation coefficients of 20% for Melan-A and 28% for influenza-specific T cells. In contrast, patients with vaccination-induced T-cell responses had significantly increased T-cell frequencies clearly exceeding physiologic fluctuations. Comparable results were obtained with ELISPOT assays. In conclusion, this approach is well suited to assess T-cell responses as biologic endpoints in clinical vaccine studies.
Collapse
Affiliation(s)
- Daniel E Speiser
- Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
The past few years have seen significant progress towards understanding the mechanisms of immune surveillance and inflammation in the nervous system. In this review, the milestones of scientific discovery in this field are discussed, and the strengths and limitations of the different ways of examining the molecular pathogenesis of neuro-inflammation examined. The review is limited to the inflammatory reactions of the central nervous system that occur in multiple sclerosis and experimental autoimmune encephalomyelitis.
Collapse
Affiliation(s)
- M Bradl
- Institute for Brain Research, Department of Neuroimmunology, Vienna, Austria.
| | | |
Collapse
|
23
|
Bercovici N, Givan AL, Waugh MG, Fisher JL, Vernel-Pauillac F, Ernstoff MS, Abastado JP, Wallace PK. Multiparameter precursor analysis of T-cell responses to antigen. J Immunol Methods 2003; 276:5-17. [PMID: 12738355 DOI: 10.1016/s0022-1759(03)00059-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Triggering of the T-cell receptor by cognate antigen induces a variety of cellular events leading to cell proliferation and differentiation. While the plasticity and diversity of T-cell responses have been recognized for a long time, few quantitative studies have been conducted to measure what proportion of specific T cells will enter a given differentiation program after antigen stimulation. In the present study, we analyzed human T cells cultured with influenza-peptide-loaded dendritic cells. We compared three individual methods for assaying the frequency of antigen-specific T cells: ELISPOT, tetramer-binding, and proliferation. The three methods yielded similar but not identical results. In order to study these differences at the single cell level, we developed a multiparameter flow cytometric method, which allows simultaneous analysis of antigen-specific tetramer binding, T-cell proliferation, and cytokine production. Based on these data, we used flow precursor frequency analysis to calculate the proportion of eight different precursor subsets in the original, resting population. We conclude that approximately half of the cells that bound specific tetramers actually proliferated and synthesized IFNgamma in response to antigen. In addition, similar numbers of cells that did not bind tetramer proliferated (but did not synthesize IFNgamma). The method allows for an estimate of the precursor frequency of each functional subset within the initial population. It could be applied to additional markers of function and differentiation, combining all parameters into a description of the complex response potential of a T-cell pool.
Collapse
|
24
|
Thayer WP, Dao CT, Ignatowicz L, Jensen PE. A novel single chain I-A(b) molecule can stimulate and stain antigen-specific T cells. Mol Immunol 2003; 39:861-70. [PMID: 12686502 DOI: 10.1016/s0161-5890(03)00010-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Multimers of soluble major histocompatibility complex class I and II molecules have proven to be useful reagents in quantifying and following specific T cell populations. This study describes the design, generation, and characterization of a novel, single chain I-A(b) molecule which utilizes a unique linker derived from the murine invariant chain. A fragment of the invariant chain, residues 58-85, binds to a region proximal to the class II peptide binding groove and stabilizes occupancy of the class II invariant chain-associated peptide. We have utilized this fragment, replacing CLIP with the Ealpha peptide sequence, to lock the attached peptide into the class II binding groove. The single chain I-A(b) molecule was recognized by a panel of conformation-sensitive, I-A(b)-specific, monoclonal antibodies. Membrane-bound and soluble forms of the single chain I-A(b) stimulated an antigen-specific T cell hybridoma, and tetramers made from soluble monomers stained these cells. The unique features of this molecule may be useful in the design of recombinant T cell receptor ligands containing peptides with low affinity for MHC.
Collapse
Affiliation(s)
- Wesley P Thayer
- Department of Pathology and Laboratory of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
25
|
Catros-Quemener V, Bouet F, Genetet N. [Antitumor immunity and cellular cancer therapies]. Med Sci (Paris) 2003; 19:43-53. [PMID: 12836191 DOI: 10.1051/medsci/200319143] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The identification of tumor specific antigens has provided important advance in tumor immunology. It is now established that specific cytotoxic T lymphocytes (CTL) and natural killer cells infiltrate tumor tissues and are effector cells able to control tumor growth. However, such a natural antitumor immunity has limited effects in cancer patients. Failure of host defenses against tumor is consecutive to several mechanisms which are becoming targets to design new immunotherapeutic approaches. CTL are critical components of the immune response to human tumors and induction of strong CTL responses is the goal of most current vaccine strategies. Effectiveness of cytokine therapy, cancer vaccines and injection of cells improving cellular immunity have been established in tumor grafted murine models. Clinical trials are underway. To day, interest is particularly focused on cell therapy: injected cells are either "ready to use" effector cells (lymphocytes) or antigen presenting cells able to induce a protective immune reaction in vivo (dendritic cells). The challenge ahead lie in the careful optimization of the most promising strategies in clinical situation.
Collapse
Affiliation(s)
- Véronique Catros-Quemener
- UPRESA Cnrs 6027, Faculté de Médecine et CHU de Rennes, 2, avenue du Pr Léon Bernard, 35043 Rennes, France.
| | | | | |
Collapse
|
26
|
Wang LX, Chen BG, Plautz GE. Adoptive immunotherapy of advanced tumors with CD62 L-selectin(low) tumor-sensitized T lymphocytes following ex vivo hyperexpansion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:3314-20. [PMID: 12218152 DOI: 10.4049/jimmunol.169.6.3314] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tumor-draining lymph nodes (TDLN) contain sensitized T cells with the phenotype CD62 L-selectin(low) (CD62L(low)) that can be activated ex vivo with anti-CD3 mAb and IL-2 to acquire potent dose-dependent effector function manifested upon adoptive transfer to secondary tumor-bearing hosts. In this study advanced tumor models were used as a stringent comparison of efficacy for the CD62L(low) subset, comprising 5-7% of the TDLN cells, vs the total population of TDLN cells following culture in high dose IL-2 (100 U/ml). During the 9-day activation period the total number of CD8+ T cells increased 1500-fold, with equivalent proliferation in the CD62L(low) vs the total TDLN cell cultures. Adoptive transfer of activated CD62L(low) cells eliminated 14-day pulmonary metastases and cured 10-day s.c. tumors, whereas transfer of maximally tolerated numbers of total TDLN cells was not therapeutic. Despite their propagation in a high concentration of IL-2, the hyperexpanded CD62L(low) subset of TDLN cells functioned in vivo without exogenous IL-2, and CD8+ T cells demonstrated relative helper independence. Moreover, the anti-tumor response was specific for the sensitizing tumor, and long term memory was established. The facile enrichment of tumor-reactive TDLN T cells, based on the CD62L(low) phenotype, circumvents the need for prior knowledge of the relevant tumor Ags. Coupling the isolation of pre-effector T cells with rapid ex vivo expansion to >3 logs could overcome some of the shortcomings of active immunotherapy or in vivo cytokine treatment, where selective robust expansion of effector cells has been difficult to achieve.
Collapse
Affiliation(s)
- Li-Xin Wang
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | |
Collapse
|
27
|
Nagorsen D, Monsurrò V, Wang E, Marincola FM. Characterization of CD8(-) HLA class I/epitope tetrameric complexes binding T cells. J Immunother 2002; 25:379-84. [PMID: 12218775 DOI: 10.1097/00002371-200209000-00001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Antigen-specific CD8-expressing T cells play a crucial role in the host's defense against viral disease and malignancy. Epitope-specific CD8(+)T cell responses to malignant and viral disease can be accurately measured using tetramers (tHLA) of HLA class I molecules loaded with antigenic peptides. In addition, tHLA have been used to evaluate immune responses to antigen-specific immunization. tHLA bind specifically to complementary T-cell receptor (TCR) structures on the surface of T cells expressing the CD8 coreceptor. Surprisingly, however, CD8(-) cells binding tHLA are often observed. This study uses four-color flow cytometry to show that HLA-A*0201-tHLA-stained CD8(-) cells can be divided into two subsets: 87% represent B-lymphocytes (CD19(+), CD45RA(+), HLA-DR(+), and CD20(+)), and 13% represent T-helper cells (CD3(+), CD4(+), CD45RA(+), and CD27(+)). This phenomenon is not HLA-restricted because it could be observed even in peripheral blood mononuclear cells (PBMC) from a non HLA-A*0201-expressing healthy donor. In addition, no T-cell receptor was detected on the B-lymphocytes. Retrospective enumeration of vaccine-induced CD8(-) tHLA cells in 243 PBMC samples from 36 patients with melanoma undergoing peptide vaccination revealed that tHLA staining is not dependent on immunization status or the presence of CD8(+) tHLA(+) T cells. These findings, suggest that the nonspecific binding of tHLA to non-TCR-expressing T cells requires a careful interpretation of results and further steps in preparation of sample for tHLA-based sorting of epitope specific T cells.
Collapse
Affiliation(s)
- Dirk Nagorsen
- Immunogenetics Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland 20982-1502, USA
| | | | | | | |
Collapse
|